1
|
Shen J, Duan X, Xie T, Zhang X, Cai Y, Pan J, Zhang X, Sun X. Advances in locally administered nucleic acid therapeutics. Bioact Mater 2025; 49:218-254. [PMID: 40144794 PMCID: PMC11938090 DOI: 10.1016/j.bioactmat.2025.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Nucleic acid drugs represent the latest generation of precision therapeutics, holding significant promise for the treatment of a wide range of intractable diseases. Delivery technology is crucial for the clinical application of nucleic acid drugs. However, extrahepatic delivery of nucleic acid drugs remains a significant challenge. Systemic administration often fails to achieve sufficient drug enrichment in target tissues. Localized administration has emerged as the predominant approach to facilitate extrahepatic delivery. While localized administration can significantly enhance drug accumulation at the injection sites, nucleic acid drugs still face biological barriers in reaching the target lesions. This review focuses on non-viral nucleic acid drug delivery techniques utilized in local administration for the treatment of extrahepatic diseases. First, the classification of nucleic acid drugs is described. Second, the current major non-viral delivery technologies for nucleic acid drugs are discussed. Third, the bio-barriers, administration approaches, and recent research advances in the local delivery of nucleic acid drugs for treating lung, brain, eye, skin, joint, and heart-related diseases are highlighted. Finally, the challenges associated with the localized therapeutic application of nucleic acid drugs are addressed.
Collapse
Affiliation(s)
- Jie Shen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xusheng Duan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Ting Xie
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xinrui Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yue Cai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Junhao Pan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xin Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuanrong Sun
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| |
Collapse
|
2
|
Ding S, Alexander E, Liang H, Kulchar RJ, Singh R, Herzog RW, Daniell H, Leong KW. Synthetic and Biogenic Materials for Oral Delivery of Biologics: From Bench to Bedside. Chem Rev 2025. [PMID: 40168474 DOI: 10.1021/acs.chemrev.4c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
The development of nucleic acid and protein drugs for oral delivery has lagged behind their production for conventional nonoral routes. Over the past decade, the evolution of DNA- and RNA-based technologies combined with the innovation of state-of-the-art delivery vehicles for nucleic acids has brought rapid advancements to the biopharmaceutical field. Nucleic acid therapies have the potential to achieve long-lasting effects, or even cures, by inhibiting or editing genes, which is not possible with conventional small-molecule drugs. However, challenges and limitations must be addressed before these therapies can provide cures for chronic conditions and rare diseases, rather than only offering temporary relief. Nucleic acids and proteins face premature degradation in the acidic, enzyme-rich stomach environment and are rapidly cleared by the liver. To overcome these challenges, various delivery vehicles have been developed to transport therapeutic compounds to the intestines, where the active compounds are released and gut microbiota and mucosal immune system also play an important role. This review provides a comprehensive overview of the promises and pitfalls associated with the oral route of administration of biologics, current delivery systems, applications of orally delivered therapeutics, and the challenges and considerations for translation of nucleic acid and protein therapeutics into clinical practice.
Collapse
Affiliation(s)
- Suwan Ding
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Elena Alexander
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Huiyi Liang
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Rachel J Kulchar
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Rahul Singh
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| |
Collapse
|
3
|
Mansour HM, El-Khatib AS. Oligonucleotide-based therapeutics for neurodegenerative disorders: Focus on antisense oligonucleotides. Eur J Pharmacol 2025; 998:177529. [PMID: 40118328 DOI: 10.1016/j.ejphar.2025.177529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 03/04/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Antisense oligonucleotides (ASOs) specifically bind to target RNA sequences and regulate protein expression through various mechanisms. ASOs are a promising therapeutic approach for treating neurodegenerative diseases. The ASO field is a growing area of drug development that focuses on targeting the root cause of diseases at the RNA level, providing a promising alternative to therapies that target downstream processes. Addressing challenges related to off-target effects and inadequate biological activity is essential to successfully develop ASO-based therapies. Researchers have investigated various chemical modifications and delivery strategies to overcome these challenges. This review discusses oligonucleotide-based therapies, particularly ASOs. We discuss the chemical modifications and mechanisms of action of ASOs. Additionally, we recap the results of preclinical and clinical studies testing different ASOs in various neurodegenerative disorders, including spinal muscular atrophy, Huntington's disease, amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease. In conclusion, ASO drugs show promise as a therapeutic option for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Heba M Mansour
- Central Administration of Biologicals, Innovative Products, and Clinical Studies, Egyptian Drug Authority, EDA, Giza, Egypt.
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
4
|
Liu Y, Wang C, Fu X, Ren M. The Progress and Evolving Trends in Nucleic-Acid-Based Therapies. Biomolecules 2025; 15:376. [PMID: 40149911 PMCID: PMC11940734 DOI: 10.3390/biom15030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Nucleic-acid-based therapies have emerged as a pivotal domain within contemporary biomedical science, marked by significant advancements in recent years. These innovative treatments primarily operate through the precise binding of DNA or RNA molecules to discrete target genes, subsequently suppressing the expression of the target proteins. The spectrum of nucleic-acid-based therapies encompasses antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs), etc. Compared to more traditional medicinal approaches, nucleic-acid-based therapies stand out for their highly targeted action on specific genes, as well as their potential for chemical modification to improve resistance to nucleases, ensuring sustained therapeutic activity and mitigating immunogenicity concerns. Nevertheless, these molecules' limited cellular permeability necessitates the deployment of delivery vectors to enhance their intracellular uptake and stability. As nucleic-acid-based therapies progressively display promising pharmacodynamic profiles, there has been a burgeoning interest in these treatments for applications in clinical research. This review aims to summarize the variety of nucleic acid drugs and their mechanisms, evaluate the present status in research and application, discourse on prospective trends, and potential challenges ahead. These innovative therapeutics are anticipated to assume a pivotal role in the management of a wide array of diseases.
Collapse
Affiliation(s)
| | | | - Xiuping Fu
- School of Chemistry and School of Life Sciences, Tiangong University, Tianjin 300387, China; (Y.L.); (C.W.)
| | - Mengtian Ren
- School of Chemistry and School of Life Sciences, Tiangong University, Tianjin 300387, China; (Y.L.); (C.W.)
| |
Collapse
|
5
|
Ma Z, Zou B, Zhao J, Zhang R, Zhu Q, Wang X, Xu L, Gao X, Hu X, Feng W, Luo W, Wang M, He Y, Yu Z, Cui W, Zhang Q, Kuai L, Su W. Development of a DNA-encoded library screening method "DEL Zipper" to empower the study of RNA-targeted chemical matter. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100204. [PMID: 39716586 DOI: 10.1016/j.slasd.2024.100204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/15/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024]
Abstract
To date, RNA-targeted chemical matter is under explored due to a lack of robust screening assays. In this study, we present a novel RNA-targeted small molecule screening approach using a specialized DNA-encoded library (DEL). Our findings reveal that the specialized DEL library, called "DEL Zipper", can significantly reduce single-stranded DNA-RNA region interaction signals during various kinds of RNA selection. By performing the selection against both G-quadruplex, we have identified novel hits that interact with RNA targets and the results are validated through binding. This study demonstrates that the "DEL Zipper" method is a robust screening assay that has potential for discovering small molecule ligands for diverse RNA targets.
Collapse
Affiliation(s)
- Zhongyao Ma
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Bin Zou
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Jiannan Zhao
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Rui Zhang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Qiaoqiao Zhu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Xiaofeng Wang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Linan Xu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Xiang Gao
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Xinyue Hu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Wei Feng
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Wen Luo
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Min Wang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Yunyun He
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Zhifeng Yu
- WuXi AppTec, 55 Cambridge Parkway, 8th Floor, Cambridge, MA 02142, United States
| | - Weiren Cui
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Qi Zhang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Letian Kuai
- WuXi AppTec, 55 Cambridge Parkway, 8th Floor, Cambridge, MA 02142, United States.
| | - Wenji Su
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China.
| |
Collapse
|
6
|
Palaiologou A, Naki M, Pantazopoulou M, Kattan FG, Stefanis L, Doxakis E, Tamvakopoulos C. A Bioanalytical Liquid Chromatography Tandem Mass Spectrometry Approach for the Quantification of a Novel Antisense Oligonucleotide Designed for Parkinson's Disease: A Rat Brain Biodistribution Study. ACS Pharmacol Transl Sci 2025; 8:592-601. [PMID: 39974630 PMCID: PMC11833717 DOI: 10.1021/acsptsci.4c00698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/21/2025]
Abstract
Antisense oligonucleotides (ASOs) represent a unique category of therapeutics targeting disease-related RNAs. Since this new therapeutic category emerged, the immediate need to analyze ASOs in clinically relevant biological matrices has led to several methodologies, such as ligand binding assays and imaging techniques. To overcome issues in specificity and provide exact quantitative data for ASOs, a new LC-MS/MS method was developed to analyze in brain tissue a novel 4-10-4 gapmer ASO with the potential for treating Parkinson's disease with phosphorothioated backbone and 2'-O-(2-methoxyethyl) modifications. The sample pretreatment protocol to extract the ASO from brain tissue employed solid phase extraction (SPE) and protein digestion. The LC-MS/MS method was fully optimized, validated and applied to quantify the target ASO in brain tissue samples following an in vivo brain distribution study. The method has a Lower Limit Of Quantification of 1 ng/mg and was applied to the study's samples, demonstrating satisfactory sensitivity and providing valuable information about the ASO's distribution in different brain regions over 45 days.
Collapse
Affiliation(s)
- Anastasia Palaiologou
- Center
of Clinical Research, Experimental Surgery and Translational Research,
Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou Street 4, Athens GR-11527, Greece
| | - Marianna Naki
- Center
of Basic Research, Biomedical Research Foundation,
Academy of Athens, 11527 Athens, Greece
- Department
of Physiology, National and Kapodistrian
University of Athens (NKUA), 11527 Athens, Greece
| | - Marina Pantazopoulou
- Center
of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou Street 4, Athens GR-11527, Greece
| | - Fedon-Giasin Kattan
- Center
of Basic Research, Biomedical Research Foundation,
Academy of Athens, 11527 Athens, Greece
| | - Leonidas Stefanis
- Center
of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou Street 4, Athens GR-11527, Greece
| | - Epaminondas Doxakis
- Center
of Basic Research, Biomedical Research Foundation,
Academy of Athens, 11527 Athens, Greece
| | - Constantin Tamvakopoulos
- Center
of Clinical Research, Experimental Surgery and Translational Research,
Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou Street 4, Athens GR-11527, Greece
| |
Collapse
|
7
|
Leckie J, Yokota T. Integrating Machine Learning-Based Approaches into the Design of ASO Therapies. Genes (Basel) 2025; 16:185. [PMID: 40004514 PMCID: PMC11855077 DOI: 10.3390/genes16020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Rare diseases impose a significant burden on affected individuals, caregivers, and healthcare systems worldwide. Developing effective therapeutics for these small patient populations presents substantial challenges. Antisense oligonucleotides (ASOs) have emerged as a promising therapeutic approach that targets the underlying genetic cause of disease at the RNA level. Several ASOs have gained FDA approval for the treatment of genetic conditions, including use in personalized N-of-1 trials. However, despite their potential, ASOs often exhibit limited clinical efficacy, and optimizing their design is a complex process influenced by numerous factors. Machine learning-based platforms, including eSkip-Finder and ASOptimizer, have been developed to address these challenges by predicting optimal ASO sequences and chemical modifications to enhance efficacy. eSkip-Finder focuses on exon-skipping applications, while ASOptimizer aims to optimize ASOs for RNA degradation. Preliminary in vitro results have demonstrated the promising predictive power of these platforms. However, limitations remain, including their generalizability to alternative targets and gaps in their consideration of all factors influencing ASO efficacy and safety. Continued advancements in machine learning models, alongside efforts to incorporate additional features affecting ASO efficacy and safety, hold significant promise for the field. These platforms have the potential to streamline ASO development, reduce associated costs, and improve clinical outcomes, positioning machine learning as a key tool in the future of ASO therapeutics.
Collapse
Affiliation(s)
- Jamie Leckie
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada;
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada;
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Sciences Research, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
8
|
Linthorst NA, van Vlijmen BJ, Eikenboom JC. The future of siRNA-mediated approaches to treat von Willebrand disease. Expert Rev Hematol 2025; 18:109-122. [PMID: 39865861 PMCID: PMC11854048 DOI: 10.1080/17474086.2025.2459259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/19/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION The clinical management of the inherited bleeding disorder von Willebrand disease (VWD) focuses on normalizing circulating levels of von Willebrand factor (VWF) and factor VIII (FVIII) to prevent or control bleeding events. The heterogeneous nature of VWD, however, complicates effective disease management and development of universal treatment guidelines. AREAS COVERED The current treatment modalities of VWD and their limitations are described and why this prompts the development of new treatment approaches. In particular, RNA-based therapeutics have gained significant interest because of their ability to reversibly alter gene expression with long-term efficacy. In the field of VWD, small-interfering RNAs (siRNAs) have been explored through various strategies to improve disease phenotypes. These different approaches are discussed as well as their potential impact on reshaping the future therapeutic landscape. EXPERT OPINION Current treatments for VWD often require frequent intravenous administration of VWF concentrates or desmopressin, with only short-term benefits. Moreover, remaining circulating mutant VWF can cause detrimental effects. Allele-selective siRNA-based therapies could provide more reliable and long-term disease correction by specifically targeting mutant VWF. This approach could be applied to a large part of the population aligning with the growing emphasis on personalized treatment and patient-centered care in VWD management.
Collapse
Affiliation(s)
- Noa A. Linthorst
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart J.M van Vlijmen
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen C.J Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
9
|
Zhou KXT, Bujold KE. The Emergence of Oligonucleotide Building Blocks in the Multispecific Proximity-Inducing Drug Toolbox of Destruction. ACS Chem Biol 2025; 20:3-18. [PMID: 39704048 DOI: 10.1021/acschembio.4c00311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Oligonucleotides are a rapidly emerging class of therapeutics. Their most well-known examples are informational drugs that modify gene expression by binding mRNA. Despite inducing proximity between biological machinery and mRNA when applied to modulating gene expression, oligonucleotides are not typically labeled as "proximity-inducing" in literature. Yet, they have recently been explored as building blocks for multispecific proximity-inducing drugs (MPIDs). MPIDs are unique because they can direct endogenous biological machinery to destroy targeted molecules and cells, in contrast to traditional drugs that inhibit only their functions. The unique mechanism of action of MPIDs has enabled the targeting of previously "undruggable" molecular entities that cannot be effectively inhibited. However, the development of MPIDs must ensure that these molecules will selectively direct a potent, destruction-based mechanism of action toward intended targets over healthy tissues to avoid causing life-threatening toxicities. Oligonucleotides have emerged as promising building blocks for the design of MPIDs because they are sequence-controlled molecules that can be rationally designed to program multispecific binding interactions. In this Review, we examine the emergence of oligonucleotide-containing MPIDs in the proximity induction space, which has been dominated by antibody and small molecule MPID modalities. Moreover, examples of oligonucleotides developed as MPID candidates in immunotherapy and protein degradation are discussed to demonstrate the utility of oligonucleotides in expanding the scope and selectivity of the MPID toolbox. Finally, we discuss the utility of programming "AND" gates into oligonucleotide scaffolds to encode conditional responses that have the potential to be incorporated into MPIDs, which can further enhance their selectivity, thus increasing the scope of this drug category.
Collapse
Affiliation(s)
- Kevin Xiao Tong Zhou
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ONL8S 4M1, Canada
| | - Katherine E Bujold
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ONL8S 4M1, Canada
| |
Collapse
|
10
|
Anand P, Zhang Y, Patil S, Kaur K. Metabolic Stability and Targeted Delivery of Oligonucleotides: Advancing RNA Therapeutics Beyond The Liver. J Med Chem 2025. [PMID: 39772535 DOI: 10.1021/acs.jmedchem.4c02528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Oligonucleotides have emerged as a formidable new class of nucleic acid therapeutics. Fully modified oligonucleotides exhibit enhanced metabolic stability and display successful clinical applicability for targets formerly considered "undruggable". Accumulating studies show that conjugation to targeting modalities of stabilized oligonucleotides, especially small interfering RNAs (siRNAs), has enabled robust delivery to intended cells/tissues. However, the major challenge in the field has been the stability and targeted delivery of oligonucleotides (siRNAs and antisense oligonucleotides (ASOs)) to extrahepatic tissues. In this Perspective, we review chemistry innovations and emerging delivery approaches that have revolutionized oligonucleotide drug discovery and development. We explore findings from both academia and industry that highlight the potential of oligonucleotides for indications involving different extrahepatic organs─including skeletal muscles, brain, lungs, skin, heart, adipose tissue, and eyes. In all, continued advances in chemistry coupled with conjugation-based approaches or novel administration routes will further advance the delivery of oligonucleotides to extrahepatic tissues.
Collapse
Affiliation(s)
- Puneet Anand
- Regeneron Genetic Medicines, Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Yu Zhang
- Regeneron Genetic Medicines, Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Spoorthi Patil
- Regeneron Genetic Medicines, Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Keerat Kaur
- Regeneron Genetic Medicines, Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| |
Collapse
|
11
|
Tzaban S, Stern O, Zisman E, Eisenberg G, Klein S, Frankenburg S, Lotem M. Alternative splicing of modulatory immune receptors in T lymphocytes: a newly identified and targetable mechanism for anticancer immunotherapy. Front Immunol 2025; 15:1490035. [PMID: 39845971 PMCID: PMC11752881 DOI: 10.3389/fimmu.2024.1490035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Abstract
Alternative splicing (AS) is a mechanism that generates translational diversity within a genome. Equally important is the dynamic adaptability of the splicing machinery, which can give preference to one isoform over others encoded by a single gene. These isoform preferences change in response to the cell's state and function. Particularly significant is the impact of physiological alternative splicing in T lymphocytes, where specific isoforms can enhance or reduce the cells' reactivity to stimuli. This process makes splicing isoforms defining features of cell states, exemplified by CD45 splice isoforms, which characterize the transition from naïve to memory states. Two developments have accelerated the use of AS dynamics for therapeutic interventions: advancements in long-read RNA sequencing and progress in nucleic acid chemical modifications. Improved oligonucleotide stability has enabled their use in directing splicing to specific sites or modifying sequences to enhance or silence particular splicing events. This review highlights immune regulatory splicing patterns with potential significance for enhancing anticancer immunotherapy.
Collapse
Affiliation(s)
- Shay Tzaban
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ori Stern
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elad Zisman
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Galit Eisenberg
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Center for Melanoma and Cancer Immunotherapy, Sharett Institute of Oncology, Jerusalem, Israel
| | - Shiri Klein
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Center for Melanoma and Cancer Immunotherapy, Sharett Institute of Oncology, Jerusalem, Israel
| | - Shoshana Frankenburg
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Lotem
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Center for Melanoma and Cancer Immunotherapy, Sharett Institute of Oncology, Jerusalem, Israel
- Hadassah Cancer Research Institute, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
12
|
Leśnikowski ZJ. Is it Time for Multi-Drug Therapy with Combination of Therapeutic Nucleic Acids? ChemMedChem 2025; 20:e202400493. [PMID: 39415702 DOI: 10.1002/cmdc.202400493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Therapeutic nucleic acids (TNAs) are a new class of drugs that exhibit different properties and mechanisms of action from those of small molecules or biological drugs. Over twenty oligonucleotide drugs and several COVID-19 vaccines have received regulatory approval for clinical use. A characteristic feature of these TNAs is that they are directed against one specific biological target and one specific RNA or DNA sequence. Consequently, TNAs currently used are administered as monotherapy. Due to the known advantages of multidrug therapy with low molecular weight drugs, it may be time to intensify work on such a treatment protocol, also in the case of TNAs.
Collapse
Affiliation(s)
- Zbigniew J Leśnikowski
- Department of Medicinal Chemistry, Institute of Medical Biology PAS, Lodowa 106, 92-232, Łódź, Poland
| |
Collapse
|
13
|
Tartaglia MC, Ingelsson M. Molecular Therapeutics in Development to Treat Alzheimer's Disease. Mol Diagn Ther 2025; 29:9-24. [PMID: 39316339 PMCID: PMC11748464 DOI: 10.1007/s40291-024-00738-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 09/25/2024]
Abstract
Until recently, only symptomatic therapies, in the form of acetylcholine esterase inhibitors and NMDA-receptor antagonists, have been available for the treatment of Alzheimer's disease. However, advancements in our understanding of the amyloid cascade hypothesis have led to a development of disease-modifying therapeutic strategies. These include immunotherapies based on an infusion of monoclonal antibodies against amyloid-β, three of which have been approved for the treatment of Alzheimer's disease in the USA (one of them, lecanemab, has also been approved in several other countries). They all lead to a dramatic reduction of amyloid plaques in the brain, whereas their clinical effects have been more limited. Moreover, they can all lead to side effects in the form of amyloid-related imaging abnormalities. Ongoing developments aim at facilitating their administration, further improving their effects and reducing the risk for amyloid-related imaging abnormalities. Moreover, a number of anti-tau immunotherapies are in clinical trials, but none has so far shown any robust effects on symptoms or pathology. Another line of development is represented by gene therapy. To date, only antisense oligonucleotides against amyloid precursor protein/amyloid-β and tau have reached the clinical trial stage but a variety of gene editing strategies, such as clustered regularly interspaced short palindromic repeats/Cas9-mediated non-homologous end joining, base editing, and prime editing, have all shown promise on preclinical disease models. In addition, a number of other pharmacological compounds targeting a multitude of biochemical processes, believed to be centrally involved in Alzheimer's disease, are currently being evaluated in clinical trials. This article delves into current and future perspectives on the treatment of Alzheimer's disease, with an emphasis on immunotherapeutic and gene therapeutic strategies.
Collapse
Affiliation(s)
- Maria Carmela Tartaglia
- Krembil Brain Institute, University Health Network, 6th Floor, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Martin Ingelsson
- Krembil Brain Institute, University Health Network, 6th Floor, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada.
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.
- Department of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
14
|
Ruchi R, Raman GM, Kumar V, Bahal R. Evolution of antisense oligonucleotides: navigating nucleic acid chemistry and delivery challenges. Expert Opin Drug Discov 2025; 20:63-80. [PMID: 39653607 PMCID: PMC11823135 DOI: 10.1080/17460441.2024.2440095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024]
Abstract
INTRODUCTION Antisense oligonucleotide (ASO) was established as a viable therapeutic option for genetic disorders. ASOs can target RNAs implicated in various diseases, including upregulated mRNA and pre-mRNA undergoing abnormal alternative splicing events. Therapeutic applications of ASOs have been proven with the Food and Drug Administration approval of several drugs in recent years. Earlier enzymatic stability and delivery remains a big challenge for ASOs. Introducing new chemical modifications and new formulations resolving the issues related to the nuclease stability and delivery of the ASOs. Excitingly, ASOs-based bioconjugates that target the hepatocyte have gained much attraction. Efforts are ongoing to increase the therapeutic application of the ASOs to the extrahepatic tissue as well. AREA COVERED We have briefly discussed the mechanism of ASOs, the development of new chemistries, and delivery strategies for ASO-based drug discovery and development. The discussion focuses more on the already approved ASOs and those in the clinical development stage. EXPERT OPINION To expand the clinical application of ASOs, continuous effort is required to develop precise delivery strategies for targeting extrahepatic tissue to minimize the off-target effects.
Collapse
Affiliation(s)
- Ruchi Ruchi
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Govind Mukesh Raman
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
- Farmington High School, Farmington, CT, USA
| | - Vikas Kumar
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
15
|
Ogieuhi IJ, Callender K, Odukudu GDO, Obi ES, Muzofa K, Babalola AE, Ugiomoh OMA, Umenzeakor KH, Akingbola A, Ayoson CO, Agbo EU, Odoeke MC. Antisense Oligonucleotides in Dyslipidemia Management: A Review of Clinical Trials. High Blood Press Cardiovasc Prev 2025; 32:33-47. [PMID: 39476283 DOI: 10.1007/s40292-024-00682-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/06/2024] [Indexed: 01/31/2025] Open
Abstract
INTRODUCTION Elevated serum total cholesterol levels, very low-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, or a decreased serum high-density lipoprotein cholesterol concentration characterize dyslipidemia. Antisense Oligonucleotide therapy in dyslipidemia targets apolipoprotein B (ApoB), an essential component of low-density lipoprotein (LDL) associated with atherosclerosis development. AIM This review aims to critically evaluate the efficacy and safety of this group of medications in mitigating dyslipidemia in at-risk individuals and its potential role in advancing personalized medicine in the management of dyslipidemias. METHODS A detailed search was conducted from multiple databases adhering to the PRISMA guidelines. Clinical trials and randomized controlled trials on antisense oligonucleotides for management of dyslipidemias were included, excluding non-English studies, case reports and all forms of reviews. Data was screened, with duplicates removed, and key findings were synthesized using a narrative approach. RESULTS AND CONCLUSION The potential of antisense oligonucleotides (ASOs) to treat dyslipidemia and other disorders has attracted much interest. Several studies and clinical trials have been conducted on the safety and tolerability of ASOs for dyslipidemia. Although statins are the mainstay management of hypercholesterolemia, there is evidence from clinical trials that ASOs can even be more effective with little to no side effects. Novel therapeutic approaches such as antisense oligonucleotides (ASOs) offer tailored therapeutic alternatives. ASOs such as Mipomersen and Volanesorsen provide additional treatment options for patients with inherited lipid abnormalities by lowering certain atherogenic lipoproteins such as apo B and ApoC-III, respectively.
Collapse
Affiliation(s)
| | - Kristen Callender
- Queen Elizabeth Hospital, Martindales Road, Bridgetown, St. Michael, Barbados
| | | | | | | | | | | | | | - Adewunmi Akingbola
- Department of Public Health, University of Cambridge, Cambridge, England, UK
| | | | | | | |
Collapse
|
16
|
Klabenkova K, Zakhryamina A, Burakova E, Bizyaev S, Fokina A, Stetsenko D. Synthesis of New Polyfluoro Oligonucleotides via Staudinger Reaction. Int J Mol Sci 2024; 26:300. [PMID: 39796153 PMCID: PMC11719919 DOI: 10.3390/ijms26010300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Nowadays, nucleic acid derivatives capable of modulating gene expression at the RNA level have gained widespread recognition as promising therapeutic agents. A suitable degree of biological stability of oligonucleotide therapeutics is required for in vivo application; this can be most expeditiously achieved by the chemical modification of the internucleotidic phosphate group, which may also affect their cellular uptake, tissue distribution and pharmacokinetics. Our group has previously developed a strategy for the chemical modification of the phosphate group via the Staudinger reaction on a solid phase of the intermediate dinucleoside phosphite triester and a range of, preferably, electron deficient organic azides such as sulfonyl azides during automated solid-phase DNA synthesis according to the conventional β-cyanoethyl phosphoramidite scheme. Polyfluoro compounds are characterized by unique properties that have prompted their extensive application both in industry and in scientific research. We report herein the synthesis and isolation of novel oligodeoxyribonucleotides incorporating internucleotidic perfluoro-1-octanesulfonyl phosphoramidate or 2,2,2-trifluoroethanesulfonyl phosphoramidate groups. In addition, novel oligonucleotide derivatives with fluorinated zwitterionic phosphate mimics were synthesized by a tandem methodology, which involved (a) the introduction of a carboxylic ester group at the internucleotidic position via the Staudinger reaction with methyl 2,2-difluoro-3-azidosulfonylacetate; and (b) treatment with an aliphatic diamine, e.g., 1,1-dimethylethylenediamine or 1,3-diaminopropane. It was further shown that the polyfluoro oligonucleotides obtained were able to form complementary duplexes with either DNA or RNA, which were not significantly differing in stability from the natural counterparts. Long-chain perfluoroalkyl oligonucleotides were taken up into cultured human cells in the absence of a transfection agent. It may be concluded that the polyfluoro oligonucleotides described here can represent a useful platform for designing oligonucleotide therapeutics.
Collapse
Affiliation(s)
- Kristina Klabenkova
- Department of Physics, Novosibirsk State University, 2 Pirogov Str., Novosibirsk 630090, Russia; (K.K.); (E.B.); (S.B.); (A.F.)
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Alyona Zakhryamina
- Department of Natural Sciences, Novosibirsk State University, 2 Pirogov Str., Novosibirsk 630090, Russia;
| | - Ekaterina Burakova
- Department of Physics, Novosibirsk State University, 2 Pirogov Str., Novosibirsk 630090, Russia; (K.K.); (E.B.); (S.B.); (A.F.)
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Sergei Bizyaev
- Department of Physics, Novosibirsk State University, 2 Pirogov Str., Novosibirsk 630090, Russia; (K.K.); (E.B.); (S.B.); (A.F.)
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Alesya Fokina
- Department of Physics, Novosibirsk State University, 2 Pirogov Str., Novosibirsk 630090, Russia; (K.K.); (E.B.); (S.B.); (A.F.)
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Dmitry Stetsenko
- Department of Physics, Novosibirsk State University, 2 Pirogov Str., Novosibirsk 630090, Russia; (K.K.); (E.B.); (S.B.); (A.F.)
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentiev Ave., Novosibirsk 630090, Russia
| |
Collapse
|
17
|
Stanciu SM, Jurcut R, Dragoi Galrinho R, Stefani C, Miricescu D, Rusu IR, Prisacariu GS, Mititelu R. From Molecular to Radionuclide and Pharmacological Aspects in Transthyretin Cardiac Amyloidosis. Int J Mol Sci 2024; 26:146. [PMID: 39796004 PMCID: PMC11719977 DOI: 10.3390/ijms26010146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Amyloidosis is a rare pathology characterized by protein deposits in various organs and tissues. Cardiac amyloidosis (CA) can be caused by various protein deposits, but transthyretin amyloidosis (ATTR) and immunoglobulin light chain (AL) are the most frequent pathologies. Protein misfolding can be induced by several factors such as oxidative stress, genetic mutations, aging, chronic inflammation, and neoplastic disorders. In ATTR cardiomyopathy (ATTR-CM), the amyloid fibrils can be found in the myocardium interstitial space and are associated with arrhythmias and heart failure. In pathological situations, the transthyretin (TTR) configuration is destroyed by proteolytic action, leading to monomers that further misfold and aggregate to form the amyloid fibrils. 99mTc-Pyrophosphate (99m-Tc-PYP), 99mTc 3,3-diphosphono-1,2-propanodicarboxylic acid (99m-Tc-DPD) and 99m-Tc hydroxy-methylene-Dyphosphonate (99m-Tc-HMDP) are used to detect myocardium amyloid deposits due to their ability to detect calcium ions that are present in the amyloid fibrils through dystrophic calcification. ATTR-CM therapy acts on different stages of the amyloidogenic process, including liver TTR synthesis, TTR tetramer destabilization, and misfolding of the monomers. The main aim of this narrative review is to present ATTR-CM, starting with molecular changes regarding the protein misfolding process and radionuclide aspects and finishing with pharmacological approaches.
Collapse
Affiliation(s)
- Silviu Marcel Stanciu
- Department of Internal Medicine and Gastroenterology, Carol Davila University of Medicine and Pharmacy, Central Military Emergency University Hospital, 010825 Bucharest, Romania;
| | - Ruxandra Jurcut
- Department of Cardiology, Carol Davila University of Medicine and Pharmacy, Institute of Cardiovascular Diseases “Prof CC Iliescu”, 022322 Bucharest, Romania;
| | - Ruxandra Dragoi Galrinho
- Department of Cardiology and Cardiovascular Surgery, University and Emergency Hospital, 050098 Bucharest, Romania
| | - Constantin Stefani
- Department I of Family Medicine and Clinical Base, “Dr. Carol Davila” Central Military Emergency University Hospital, 010825 Bucharest, Romania;
| | - Daniela Miricescu
- Discipline of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ioana Ruxandra Rusu
- Discipline of Anatomy, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Georgiana Sabina Prisacariu
- Clinic of Nuclear Medicine Central University Emergency Military Hospital “Dr Carol Davila”, 10825 Bucharest, Romania; (G.S.P.); (R.M.)
| | - Raluca Mititelu
- Clinic of Nuclear Medicine Central University Emergency Military Hospital “Dr Carol Davila”, 10825 Bucharest, Romania; (G.S.P.); (R.M.)
- Department of Nuclear Medicine, University of Medicine and Pharmacy Carol Davila, 030147 Bucharest, Romania
| |
Collapse
|
18
|
Oberemok VV, Puzanova YV, Gal’chinsky NV. The 'genetic zipper' method offers a cost-effective solution for aphid control. FRONTIERS IN INSECT SCIENCE 2024; 4:1467221. [PMID: 39726916 PMCID: PMC11670321 DOI: 10.3389/finsc.2024.1467221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/11/2024] [Indexed: 12/28/2024]
Abstract
Twenty years ago, it was difficult to imagine the use of nucleic acids in plant protection as insecticides, but today it is a reality. New technologies often work inefficiently and are very expensive; however, qualitative changes occur during their development, making them more accessible and work effectively. Invented in 2008, contact oligonucleotide insecticides (olinscides, or DNA insecticides) based on the CUAD (contact unmodified antisense DNA) platform have been substantially improved and rethought. The main paradigm shift was demonstrating that unmodified antisense DNA can act as a contact insecticide. Key breakthroughs included identifying convenient target genes (rRNA genes), mechanism of action (DNA containment), and discovering insect pests (sternorrhynchans) with high susceptibility to olinscides. Today, the CUAD platform possesses impressive characteristics: low carbon footprint, high safety for non-target organisms, rapid biodegradability, and avoidance of target-site resistance. This next-generation class of insecticides creates opportunities for developing products tailored for specific insect pest populations. The 'genetic zipper' method, based on CUAD biotechnology, integrates molecular genetics, bioinformatics, and in vitro nucleic acid synthesis. It serves as a simple and flexible tool for DNA-programmable plant protection using unmodified antisense oligonucleotides targeting pest rRNAs. Aphids, key pests of important agricultural crops, can be effectively controlled by oligonucleotide insecticides at an affordable price, ensuring efficient control with minimal environmental risks. In this article, a low-dose concentration (0.1 ng/µL; 20 mg per hectare in 200 L of water) of the 11 nt long oligonucleotide insecticide Schip-11 shows effectiveness against the aphid Schizolachnus pineti, causing mortality rate of 76.06 ± 7.68 on the 12th day (p<0.05). At a consumption rate of 200 L per hectare, the cost of the required oligonucleotide insecticide is about 0.5 USD/ha using liquid-phase DNA synthesis making them competitive in the market and very affordable for lab investigations. We also show that non-canonical base pairing Golinscide: UrRNA is well tolerated in aphids. Thus, non-canonical base-pairing should be considered not to harm non-target organisms and can be easily solved during the design of oligonucleotide insecticides. The 'genetic zipper' method, based on CUAD biotechnology, helps quickly create a plethora of efficient oligonucleotide pesticides against aphids and other pests. Already today, according to our estimations, the 'genetic zipper' is potentially capable of effectively controlling 10-15% of all insect pests using a simple and flexible algorithm.
Collapse
Affiliation(s)
- Vol V. Oberemok
- Department of General Biology and Genetics, Institute of Biochemical Technologies, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, Simferopol, Republic of Crimea
- Laboratory of Entomology and Phytopathology, Dendrology and Landscape Architecture, Nikita Botanical Gardens—National Scientific Centre of the Russian Academy of Sciences, Yalta, Republic of Crimea
| | - Yelizaveta V. Puzanova
- Department of General Biology and Genetics, Institute of Biochemical Technologies, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, Simferopol, Republic of Crimea
| | - Nikita V. Gal’chinsky
- Department of General Biology and Genetics, Institute of Biochemical Technologies, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, Simferopol, Republic of Crimea
| |
Collapse
|
19
|
Mikutis S, Bernardes GJL. Technologies for Targeted RNA Degradation and Induced RNA Decay. Chem Rev 2024; 124:13301-13330. [PMID: 39499674 PMCID: PMC11638902 DOI: 10.1021/acs.chemrev.4c00472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/03/2024] [Accepted: 10/29/2024] [Indexed: 11/07/2024]
Abstract
The vast majority of the human genome codes for RNA, but RNA-targeting therapeutics account for a small fraction of approved drugs. As such, there is great incentive to improve old and develop new approaches to RNA targeting. For many RNA targeting modalities, just binding is not sufficient to exert a therapeutic effect; thus, targeted RNA degradation and induced decay emerged as powerful approaches with a pronounced biological effect. This review covers the origins and advanced use cases of targeted RNA degrader technologies grouped by the nature of the targeting modality as well as by the mode of degradation. It covers both well-established methods and clinically successful platforms such as RNA interference, as well as emerging approaches such as recruitment of RNA quality control machinery, CRISPR, and direct targeted RNA degradation. We also share our thoughts on the biggest hurdles in this field, as well as possible ways to overcome them.
Collapse
Affiliation(s)
- Sigitas Mikutis
- Yusuf Hamied Department of
Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Gonçalo J. L. Bernardes
- Yusuf Hamied Department of
Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| |
Collapse
|
20
|
Alnefaie GO. A review of the complex interplay between chemoresistance and lncRNAs in lung cancer. J Transl Med 2024; 22:1109. [PMID: 39639388 PMCID: PMC11619437 DOI: 10.1186/s12967-024-05877-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
Lung Cancer (LC) is characterized by chemoresistance, which poses a significant clinical challenge and results in a poor prognosis for patients. Long non-coding RNAs (lncRNAs) have recently gained recognition as crucial mediators of chemoresistance in LC. Through the regulation of key cellular processes, these molecules play important roles in the progression of LC and response to therapy. The mechanisms by which lncRNAs affect chemoresistance include the modulation of gene expression, chromatin structure, microRNA interactions, and signaling pathways. Exosomes have emerged as key mediators of lncRNA-driven chemoresistance, facilitating the transfer of resistance-associated lncRNAs between cancer cells and contributing to tumor development. Consequently, exosomal lncRNAs may serve as biomarkers and therapeutic targets for the treatment of LC. Therapeutic strategies targeting lncRNAs offer novel approaches to circumvent chemoresistance. Different approaches, including RNA interference (RNAi) and antisense oligonucleotides (ASOs), are available to degrade lncRNAs or alter their function. ASO-based therapies are effective at reducing lncRNA expression levels, increasing chemotherapy sensitivity, and improving clinical outcomes. The use of these strategies can facilitate the development of targeted interventions designed to disrupt lncRNA-mediated mechanisms of chemoresistance. An important aspect of this review is the discussion of the complex relationship between lncRNAs and drug resistance in LC, particularly through exosomal pathways, and the development of innovative therapeutic strategies to enhance drug efficacy by targeting lncRNAs. The development of new pathways and interventions for treating LC holds promise in overcoming this resistance.
Collapse
Affiliation(s)
- Ghaliah Obaid Alnefaie
- Department of Pathology, College of Medicine, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| |
Collapse
|
21
|
Lou W, Zhang L, Wang J. Current status of nucleic acid therapy and its new progress in cancer treatment. Int Immunopharmacol 2024; 142:113157. [PMID: 39288629 DOI: 10.1016/j.intimp.2024.113157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/05/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Nucleic acid is an essential biopolymer in all living cells, performing the functions of storing and transmitting genetic information and synthesizing protein. In recent decades, with the progress of science and biotechnology and the continuous exploration of the functions performed by nucleic acid, more and more studies have confirmed that nucleic acid therapy for living organisms has great medical therapeutic potential. Nucleic acid drugs began to become independent therapeutic agents. As a new therapeutic method, nucleic acid therapy plays an important role in the treatment of genetic diseases, viral infections and cancers. There are currently 19 nucleic acid drugs approved by the Food and Drug Administration (FDA). In the following review, we start from principles and advantages of nucleic acid therapy, and briefly describe development history of nucleic acid drugs. And then we give examples of various RNA therapeutic drugs, including antisense oligonucleotides (ASO), mRNA vaccines, small interfering RNA (siRNA) and microRNA (miRNA), aptamers, and small activating RNA (saRNA). In addition, we also focused on the current status of nucleic acid drugs used in cancer therapy and the breakthrough in recent years. Clinical trials of nucleic acid drugs for cancer treatment are under way, conventional radiotherapy and chemotherapy combined with the immunotherapies such as checkpoint inhibitors and nucleic acid drugs may be the main prospects for successful cancer treatment.
Collapse
Affiliation(s)
- Wenting Lou
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Leqi Zhang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China; Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou 310009, China.
| |
Collapse
|
22
|
Galindo-Murillo R, Cohen J, Akabayov B. Comparative molecular dynamics calculations of duplexation of chemically modified analogs of DNA used for antisense applications. NAR Genom Bioinform 2024; 6:lqae155. [PMID: 39633726 PMCID: PMC11616695 DOI: 10.1093/nargab/lqae155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 09/05/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
We have subjected several analogs of DNA that have been widely used as antisense oligonucleotide (ASO) inhibitors of gene expression to comparative molecular dynamics (MD) calculations of their ability to form duplexes with DNA and RNA. The analogs included in this study are the phosphorothioate (PS), peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleic acid (PMO), the 2'-OMe, 2'-F, 2'-methoxyethyl (2'-MOE) and the constrained cET analogs, as well as the natural phosphodiester (PO) as control, for a total of nine structures, in both XNA-DNA and XNA-RNA duplexes. This is intended as an objective criterion for their relative ability to duplex with an RNA complement and their comparative potential for antisense applications. We have found that the constrained furanose ring analogs show increased stability when considering this study's structural and energetic parameters. The 2'-MOE modification, even though energetically stable, has an elevated dynamic range and breathing properties due to the bulkier moiety in the C2' position of the furanose. The smaller modifications in the C2' position, 2'-F, 2'-OMe and PS also form stable and energetically favored duplexes with both DNA and RNA. The morpholino moiety allows for increased tolerance in accommodating either DNA or RNA and the PNA, with the PNA being the most energetically stable, although with a preference for the B-form DNA. In summary, we can rank the overall preference of hybrid strand formations as PNA > cET/LNA > PS/2'-F/2'-OMe > morpholino > 2'-MOE for the efficacy of duplex formation.
Collapse
Affiliation(s)
- Rodrigo Galindo-Murillo
- Department of Medicinal Chemistry, Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Jack S Cohen
- Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva, 8410501, Israel
| | - Barak Akabayov
- Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva, 8410501, Israel
| |
Collapse
|
23
|
Sun X, Setrerrahmane S, Li C, Hu J, Xu H. Nucleic acid drugs: recent progress and future perspectives. Signal Transduct Target Ther 2024; 9:316. [PMID: 39609384 PMCID: PMC11604671 DOI: 10.1038/s41392-024-02035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 09/20/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
High efficacy, selectivity and cellular targeting of therapeutic agents has been an active area of investigation for decades. Currently, most clinically approved therapeutics are small molecules or protein/antibody biologics. Targeted action of small molecule drugs remains a challenge in medicine. In addition, many diseases are considered 'undruggable' using standard biomacromolecules. Many of these challenges however, can be addressed using nucleic therapeutics. Nucleic acid drugs (NADs) are a new generation of gene-editing modalities characterized by their high efficiency and rapid development, which have become an active research topic in new drug development field. However, many factors, including their low stability, short half-life, high immunogenicity, tissue targeting, cellular uptake, and endosomal escape, hamper the delivery and clinical application of NADs. Scientists have used chemical modification techniques to improve the physicochemical properties of NADs. In contrast, modified NADs typically require carriers to enter target cells and reach specific intracellular locations. Multiple delivery approaches have been developed to effectively improve intracellular delivery and the in vivo bioavailability of NADs. Several NADs have entered the clinical trial recently, and some have been approved for therapeutic use in different fields. This review summarizes NADs development and evolution and introduces NADs classifications and general delivery strategies, highlighting their success in clinical applications. Additionally, this review discusses the limitations and potential future applications of NADs as gene therapy candidates.
Collapse
Affiliation(s)
- Xiaoyi Sun
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | | | - Chencheng Li
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Jialiang Hu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Hanmei Xu
- Jiangsu Province Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
24
|
Niu M, Yi W, Dong Z, Li X, Dong X, Yu L, Han Y, Zhang O, Sheng Z, An J, Li H, Sun Y. Effective inhibition of dengue virus replication using 3'UTR-targeted Vivo-Morpholinos. Front Immunol 2024; 15:1491230. [PMID: 39676860 PMCID: PMC11638040 DOI: 10.3389/fimmu.2024.1491230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/07/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Due to the impact of antibody-dependent enhancement and viral variation, effective vaccines or antiviral therapies remain lacking for the dengue virus (DENV). Nucleic acid drugs, particularly Vivo-Morpholinos (MOs), have emerged as a promising avenue for antiviral treatment due to their programmability and precise targeting, as well as their safety and stability. Method In this study, we designed and developed 10 morpho-modified (octa-guanidine dendrimer) vivo-MO molecules that target each coding gene of DENV. Subsequently, we assessed the inhibitory impact of vivo-MOs on dengue viral RNA load utilizing qRT-PCR. Furthermore, we examined the inhibitory effect on the live virus through a plaque assay and the TCID50 assay. Results We found that the vivo-3'UTR molecule targeting the 3' untranslated region of the dengue virus exhibited the highest inhibitory rate against viral load. The vivo-3'UTR demonstrated 99% inhibition of dengue virus RNA and the inhibition of up to 98% of the live virus. Additionally, the targeted sequence was conserved among all four DENV serotypes, and treatment with 10 μM of vivo-3'UTR resulted in a reduction of viral titers for all four DENV serotypes by over 99.99%. Additionally, we revealed that pre-treatment with vivo-3'UTR had a notable preventive effect against viral infection. Conclusion This study screened an effective vivo-MO target drug for the treatment of dengue virus infection, demonstrating low toxicity in mammalian cell lines, and proposed a novel preventive antiviral approach.
Collapse
Affiliation(s)
- Mengwei Niu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| | - Wenyanbo Yi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| | - Zhuofan Dong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| | - Xiaofeng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| | - Xue Dong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| | - Lifang Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| | - Yao Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| | - Oujia Zhang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ziyang Sheng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| | - Yansong Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Pals MJ, Lindberg A, Velema WA. Chemical strategies for antisense antibiotics. Chem Soc Rev 2024; 53:11303-11320. [PMID: 39436264 PMCID: PMC11495246 DOI: 10.1039/d4cs00238e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Indexed: 10/23/2024]
Abstract
Antibacterial resistance is a severe threat to modern medicine and human health. To stay ahead of constantly-evolving bacteria we need to expand our arsenal of effective antibiotics. As such, antisense therapy is an attractive approach. The programmability allows to in principle target any RNA sequence within bacteria, enabling tremendous selectivity. In this Tutorial Review we provide guidelines for devising effective antibacterial antisense agents and offer a concise perspective for future research. We will review the chemical architectures of antibacterial antisense agents with a special focus on the delivery and target selection for successful antisense design. This Tutorial Review will strive to serve as an essential guide for antibacterial antisense technology development.
Collapse
Affiliation(s)
- Mathijs J Pals
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Alexander Lindberg
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Willem A Velema
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Ghosh A, Banerjee A, Gupta S, Sinha S. A Unified Phosphoramidite Platform for the Synthesis of Morpholino Oligonucleotides and Diverse Chimeric Backbones. J Am Chem Soc 2024. [PMID: 39561294 DOI: 10.1021/jacs.4c09510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Phosphorodiamidate Morpholino Oligonucleotides (PMOs) have been well established in the milieu of FDA-approved oligonucleotide-based drugs in the past decade. Given their relevance in antisense therapeutics, a DNA/RNA synthesizer-compatible modular synthesis protocol of PMOs is long awaited to explore next-generation PMO chimeras with other therapeutically proven oligonucleotide backbones. Herein, we demonstrate a streamlined 5' → 3'phosphoramidite approach for the synthesis of PMOs using tert-butyl-protected 5'-tBu-morpholino phosphoramidites, which were synthesized from 5'-OH morpholino monomers derived from commercially available ribonucleosides. 2-Cyanoethyl (CE)-protected 5'-CE-morpholino phosphoramidites were also synthesized to generate thiophosphoramidate (TMO) and phosphoramidate (MO) morpholino oligonucleotides. Full-length PMOs and TMOs in exceptional overall yields were obtained with operational simplicity and compatibility with automated DNA/RNA synthesizers utilizing controlled pore glass (CPG) as the solid support and CH3CN as the solvent. Importantly, this method has opened the possibility of designing various biologically relevant ASO design modalities, such as PMO-TMO and PMO-MO, which were inaccessible otherwise. Moreover, DNA nucleotides were also incorporated to generate PMO-psDNA and PMO-DNA using commercially available 5'-DNA phosphoramidites. The biophysical properties of all synthesized oligonucleotides were analyzed using UV melting and circular dichroism studies. The serum interaction profile and innate immune response of the PS-modified polythymidine oligonucleotides were analyzed and it was found that the mixed backbone oligonucleotides had a balanced profile compared to fully charged or neutral extremities. Overall, the synthesis is amenable to fast generation of various types of PMO chimeras for biological screening, which will expedite their therapeutic exploration and transition to clinic.
Collapse
Affiliation(s)
- Atanu Ghosh
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Arpan Banerjee
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Shalini Gupta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Surajit Sinha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| |
Collapse
|
27
|
Tani H. Recent Advances and Prospects in RNA Drug Development. Int J Mol Sci 2024; 25:12284. [PMID: 39596348 PMCID: PMC11594839 DOI: 10.3390/ijms252212284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
RNA therapeutics have undergone remarkable evolution since their inception in the late 1970s, revolutionizing medicine by offering new possibilities for treating previously intractable diseases. The field encompasses various modalities, including antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs), each with unique mechanisms and applications. The foundation was laid in 1978 with the discovery that synthetic oligonucleotides could inhibit viral replication, followed by pivotal developments such as RNA interference's discovery in 1998. The COVID-19 pandemic marked a crucial turning point, demonstrating the potential of mRNA vaccines and accelerating interest in RNA-based approaches. However, significant challenges remain, including stability issues, delivery to target tissues, potential off-target effects, and immunogenicity concerns. Recent advancements in chemical modifications, delivery systems, and the integration of AI technologies are addressing these challenges. The field has seen notable successes, such as approved treatments for spinal muscular atrophy and hereditary transthyretin-mediated amyloidosis. Looking ahead, RNA therapeutics show promise for personalized medicine approaches, particularly in treating genetic disorders and cancer. The continued evolution of this field, driven by technological innovations and deeper understanding of RNA biology, suggests a transformative impact on future medical treatments. The purpose of this review is to provide a comprehensive overview of the evolution, current state, and prospects of RNA therapeutics.
Collapse
Affiliation(s)
- Hidenori Tani
- Department of Health Pharmacy, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama 245-0066, Japan
| |
Collapse
|
28
|
Khuu A, Verreault M, Colin P, Tran H, Idbaih A. Clinical Applications of Antisense Oligonucleotides in Cancer: A Focus on Glioblastoma. Cells 2024; 13:1869. [PMID: 39594617 PMCID: PMC11592788 DOI: 10.3390/cells13221869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Antisense oligonucleotides (ASOs) are promising drugs capable of modulating the protein expression of virtually any target with high specificity and high affinity through complementary base pairing. However, this requires a deep understanding of the target sequence and significant effort in designing the correct complementary drug. In addition, ASOs have been demonstrated to be well tolerated during their clinical use. Indeed, they are already used in many diseases due to pathogenic RNAs of known sequences and in several neurodegenerative diseases and metabolic diseases, for which they were given marketing authorizations (MAs) in Europe and the United States. Their use in oncology is gaining momentum with several identified targets, promising preclinical and clinical results, and recent market authorizations in the US. However, many challenges remain for their clinical use in cancer. It seems necessary to take a step back and review our knowledge of ASOs and their therapeutic uses in oncology. The objectives of this review are (i) to summarize the current state of the art of ASOs; (ii) to discuss the therapeutic use of ASOs in cancer; and (iii) to focus on ASO usage in glioblastoma, the challenges, and the perspective ahead.
Collapse
Affiliation(s)
- Alexandre Khuu
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Maïté Verreault
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| | - Philippe Colin
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Helene Tran
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Ahmed Idbaih
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| |
Collapse
|
29
|
Zhang X, Chen Y, Zhou S, Liu Y, Zhu S, Jia X, Lu Z, Zhang Y, Zhang W, Ye Z, Cai B, Kong L, Liu F. RNA Coating Promotes Peri-Implant Osseointegration. ACS Biomater Sci Eng 2024; 10:7030-7042. [PMID: 38943625 PMCID: PMC11558559 DOI: 10.1021/acsbiomaterials.4c00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
In addition to transmitting and carrying genetic information, RNA plays an important abiotic role in the world of nanomaterials. RNA is a natural polyanionic biomacromolecule, and its ability to promote osteogenesis by binding with other inorganic materials as an osteogenic induction agent was discovered only recently. However, whether it can promote osseointegration on implants has not been reported. Here, we investigated the effect of the RNA-containing coating materials on peri-implant osseointegration. Total RNA extracted from rat muscle tissue was used as an osteogenic induction agent, and hyaluronic acid (HA) was used to maintain its negative charge. In simulated body fluids (SBF), in vitro studies demonstrated that the resulting material encouraged calcium salt deposition. Cytological experiments showed that the RNA-containing coating induced greater cell adhesion and osteogenic differentiation in comparison to the control. The results of animal experiments showed that the RNA-containing coating had osteoinductive and bone conduction activities, which are beneficial for bone formation and osseointegration. Therefore, the RNA-containing coatings are useful for the surface modification of titanium implants to promote osseointegration.
Collapse
Affiliation(s)
- Xiao Zhang
- College
of Life Sciences, Northwest University, Xi’an 710069, China
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Yicheng Chen
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Shanluo Zhou
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Ya Liu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Simin Zhu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Xuelian Jia
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Zihan Lu
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Yufan Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Wenhui Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Zhou Ye
- Applied
Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, S.A.R., China
| | - Bolei Cai
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Liang Kong
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Fuwei Liu
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| |
Collapse
|
30
|
Pan Z, Zaman MA, Kalsoom S, Zhang Y. Messenger interference RNA therapies targeting apolipoprotein C-III and angiopoietin-like protein 3 for mixed hyperlipidemia: the future of plozasiran and zodasiran. Expert Rev Clin Pharmacol 2024; 17:1017-1023. [PMID: 39469883 DOI: 10.1080/17512433.2024.2423724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 10/30/2024]
Abstract
INTRODUCTION Mixed hyperlipidemia represents a substantial public health issue and a considerable burden on healthcare systems. Although the introduction of statins and LDL-cholesterol lowering agents have significantly reduced the incidence of atherosclerotic cardiovascular diseases (ASCVD), a significant portion of the population continues to exhibit ASCVD progression due to elevated triglyceride-rich lipoprotein (TRL) levels. This persistent risk has catalyzed the development of novel pharmacological interventions targeting these lipoproteins. AREAS COVERED Our special report commenced with a targeted PubMed search using keywords such as 'plozasiran,' 'zodasiran,' and terms related to APOC3 and ANGPTL3. As the review progressed, emergent research questions guided further searches, allowing for the inclusion of additional relevant articles to comprehensively illustrate the linkage between TRLs and cardiovascular disease, discuss the roles of APOC3, ANGPTL3, and the pharmaceutical agents that target these proteins, and provide a comparison on the ARCHES-2 and MUIR trials. EXPERT OPINION The ARCHES-2 and MUIR trials demonstrated effective triglyceride reduction by these therapies, yet it is uncertain if this correlates with significant clinical benefits. Advances in antisense oligonucleotide technology, especially the GalNAc delivery platform, show promise for personalized lipid management, though challenges such as cost and safety concerns remain.
Collapse
Affiliation(s)
- Zonghao Pan
- Department of Internal Medicine, Conemaugh Health System, Johnstown, PA, USA
| | | | - Sidra Kalsoom
- Department of Cardiology, Mercy Saint Vincent Medical Center, Toledo, OH, USA
| | - Yani Zhang
- Department of Internal Medicine, MedStar Washington Hospital Center, Washington, DC, USA
| |
Collapse
|
31
|
Jiao J, Qian Y, Lv Y, Wei W, Long Y, Guo X, Buerliesi A, Ye J, Han H, Li J, Zhu Y, Zhang W. Overcoming limitations and advancing the therapeutic potential of antibody-oligonucleotide conjugates (AOCs): Current status and future perspectives. Pharmacol Res 2024; 209:107469. [PMID: 39433169 DOI: 10.1016/j.phrs.2024.107469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
As cancer incidence rises due to an aging population, the importance of precision medicine continues to grow. Antibody-drug conjugates (ADCs) exemplify targeted therapies by delivering cytotoxic agents to specific antigens. Building on this concept, researchers have developed antibody-oligonucleotide conjugates (AOCs), which combine antibodies with oligonucleotides to regulate gene expression. This review highlights the mechanism of AOCs, emphasizing their unique ability to selectively target and modulate disease-causing proteins. It also explores the components of AOCs and their application in tumor therapy while addressing key challenges such as manufacturing complexities, endosomal escape, and immune response. The article underscores the significance of AOCs in precision oncology and discusses future directions, highlighting their potential in treating cancers driven by genetic mutations and abnormal protein expression.
Collapse
Affiliation(s)
- Jinlan Jiao
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Yun Qian
- Dermatologic Surgery Department, Institute of Dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing 210042, China
| | - Yinhua Lv
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Wenqian Wei
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Yongxuan Long
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaoling Guo
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Anya Buerliesi
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Jiahui Ye
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Hao Han
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jinbo Li
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China.
| | - Yun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, China.
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
32
|
Chaudhary N, Newby AN, Whitehead KA. Non-Viral RNA Delivery During Pregnancy: Opportunities and Challenges. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306134. [PMID: 38145340 PMCID: PMC11196389 DOI: 10.1002/smll.202306134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/25/2023] [Indexed: 12/26/2023]
Abstract
During pregnancy, the risk of maternal and fetal adversities increases due to physiological changes, genetic predispositions, environmental factors, and infections. Unfortunately, treatment options are severely limited because many essential interventions are unsafe, inaccessible, or lacking in sufficient scientific data to support their use. One potential solution to this challenge may lie in emerging RNA therapeutics for gene therapy, protein replacement, maternal vaccination, fetal gene editing, and other prenatal treatment applications. In this review, the current landscape of RNA platforms and non-viral RNA delivery technologies that are under active development for administration during pregnancy is explored. Advancements of pregnancy-specific RNA drugs against SARS-CoV-2, Zika, influenza, preeclampsia, and for in-utero gene editing are discussed. Finally, this study highlights bottlenecks that are impeding translation efforts of RNA therapies, including the lack of accurate cell-based and animal models of human pregnancy and concerns related to toxicity and immunogenicity during pregnancy. Overcoming these challenges will facilitate the rapid development of this new class of pregnancy-safe drugs.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Alexandra N. Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
33
|
Smith TD, Riedl MA. The future of therapeutic options for hereditary angioedema. Ann Allergy Asthma Immunol 2024; 133:380-390. [PMID: 38679158 DOI: 10.1016/j.anai.2024.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
Hereditary angioedema (HAE) is a rare genetic condition causing unpredictable and severe episodes of angioedema that are debilitating and life-threatening. Moreover, HAE can be classified into HAE due to C1-esterase inhibitor deficiency (HAE-C1INH) or HAE with normal C1INH. Moreover, HAE-C1INH is subcategorized as types I and II based on deficient or dysfunctional circulating C1INH protein resulting from inherited or spontaneous mutations in the SERPING1 gene leading to uncontrolled factor XII/plasma kallikrein activation and excessive bradykinin production. Bradykinin-2 receptor activation leads to vasodilation, increased vascular permeability, and smooth muscle contractions, resulting in subcutaneous or submucosal fluid extravasation that can affect the face, extremities, airway, and gastrointestinal and genitourinary systems. Furthermore, HAE with normal C1INH is caused by either a known or unknown genetic mutation, and the mechanisms are less well-established but most forms are thought to be related to bradykinin signaling with a similar presentation as HAE-C1INH despite normal levels of C1INH protein and function. Current HAE management strategies include on-demand and prophylactic treatments which replace C1INH, reduce kallikrein activity, or block bradykinin binding to the bradykinin B2 receptor. With the advent of additional small molecule inhibitors, monoclonal antibodies, RNA-targeted therapies, gene therapies, and gene modification approaches, preclinical studies and human clinical trials are underway to further expand therapeutic options in HAE. This review article will briefly summarize current HAE treatments and provide an overview of potential future therapies for HAE.
Collapse
Affiliation(s)
- Tukisa D Smith
- Division of Allergy, and Immunology, University of California, San Diego, La Jolla, California
| | - Marc A Riedl
- Division of Allergy, and Immunology, University of California, San Diego, La Jolla, California.
| |
Collapse
|
34
|
McDowall S, Aung-Htut M, Wilton S, Li D. Antisense oligonucleotides and their applications in rare neurological diseases. Front Neurosci 2024; 18:1414658. [PMID: 39376536 PMCID: PMC11456401 DOI: 10.3389/fnins.2024.1414658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/20/2024] [Indexed: 10/09/2024] Open
Abstract
Rare diseases affect almost 500 million people globally, predominantly impacting children and often leading to significantly impaired quality of life and high treatment costs. While significant contributions have been made to develop effective treatments for those with rare diseases, more rapid drug discovery strategies are needed. Therapeutic antisense oligonucleotides can modulate target gene expression with high specificity through various mechanisms determined by base sequences and chemical modifications; and have shown efficacy in clinical trials for a few rare neurological conditions. Therefore, this review will focus on the applications of antisense oligonucleotides, in particular splice-switching antisense oligomers as promising therapeutics for rare neurological diseases, with key examples of Duchenne muscular dystrophy and spinal muscular atrophy. Challenges and future perspectives in developing antisense therapeutics for rare conditions including target discovery, antisense chemical modifications, animal models for therapeutic validations, and clinical trial designs will also be briefly discussed.
Collapse
Affiliation(s)
- Simon McDowall
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
| | - May Aung-Htut
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Steve Wilton
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Dunhui Li
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| |
Collapse
|
35
|
Bubenik JL, Scotti MM, Swanson MS. Therapeutic targeting of RNA for neurological and neuromuscular disease. Genes Dev 2024; 38:698-717. [PMID: 39142832 PMCID: PMC11444190 DOI: 10.1101/gad.351612.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Neurological and neuromuscular diseases resulting from familial, sporadic, or de novo mutations have devasting personal, familial, and societal impacts. As the initial product of DNA transcription, RNA transcripts and their associated ribonucleoprotein complexes provide attractive targets for modulation by increasing wild-type or blocking mutant allele expression, thus relieving downstream pathological consequences. Therefore, it is unsurprising that many existing and under-development therapeutics have focused on targeting disease-associated RNA transcripts as a frontline drug strategy for these genetic disorders. This review focuses on the current range of RNA targeting modalities using examples of both dominant and recessive neurological and neuromuscular diseases.
Collapse
Affiliation(s)
- Jodi L Bubenik
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, the Genetics Institute, University of Florida, Gainesville, Florida 32610, USA
| | - Marina M Scotti
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, the Genetics Institute, University of Florida, Gainesville, Florida 32610, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, the Genetics Institute, University of Florida, Gainesville, Florida 32610, USA
| |
Collapse
|
36
|
Eftekhari Z, Zohrabi H, Oghalaie A, Ebrahimi T, Shariati FS, Behdani M, Kazemi-Lomedasht F. Advancements and challenges in mRNA and ribonucleoprotein-based therapies: From delivery systems to clinical applications. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102313. [PMID: 39281702 PMCID: PMC11402252 DOI: 10.1016/j.omtn.2024.102313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
The use of mRNA and ribonucleoproteins (RNPs) as therapeutic agents is a promising strategy for treating diseases such as cancer and infectious diseases. This review provides recent advancements and challenges in mRNA- and RNP-based therapies, focusing on delivery systems such as lipid nanoparticles (LNPs), which ensure efficient delivery to target cells. Strategies such as microfluidic devices are employed to prepare LNPs loaded with mRNA and RNPs, demonstrating effective genome editing and protein expression in vitro and in vivo. These applications extend to cancer treatment and infectious disease management, with promising results in genome editing for cancer therapy using LNPs encapsulating Cas9 mRNA and single-guide RNA. In addition, tissue-specific targeting strategies offer potential for improved therapeutic outcomes and reduced off-target effects. Despite progress, challenges such as optimizing delivery efficiency and targeting remain. Future research should enhance delivery efficiency, explore tissue-specific targeting, investigate combination therapies, and advance clinical translation. In conclusion, mRNA- and RNP-based therapies offer a promising avenue for treating various diseases and have the potential to revolutionize medicine, providing new hope for patients worldwide.
Collapse
Affiliation(s)
- Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Horieh Zohrabi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Tahereh Ebrahimi
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Sadat Shariati
- Department of Influenza and other Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| |
Collapse
|
37
|
Dume B, Licarete E, Banciu M. Advancing cancer treatments: The role of oligonucleotide-based therapies in driving progress. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102256. [PMID: 39045515 PMCID: PMC11264197 DOI: 10.1016/j.omtn.2024.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Although recent advancements in cancer immunology have resulted in the approval of numerous immunotherapies, minimal progress has been observed in addressing hard-to-treat cancers. In this context, therapeutic oligonucleotides, including interfering RNAs, antisense oligonucleotides, aptamers, and DNAzymes, have gained a central role in cancer therapeutic approaches due to their capacity to regulate gene expression and protein function with reduced toxicity compared with conventional chemotherapeutics. Nevertheless, systemic administration of naked oligonucleotides faces many extra- and intracellular challenges that can be overcome by using effective delivery systems. Thus, viral and non-viral carriers can improve oligonucleotide stability and intracellular uptake, enhance tumor accumulation, and increase the probability of endosomal escape while minimizing other adverse effects. Therefore, gaining more insight into fundamental mechanisms of actions of various oligonucleotides and the challenges posed by naked oligonucleotide administration, this article provides a comprehensive review of the recent progress on oligonucleotide delivery systems and an overview of completed and ongoing cancer clinical trials that can shape future oncological treatments.
Collapse
Affiliation(s)
- Bogdan Dume
- Doctoral School in Integrative Biology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| |
Collapse
|
38
|
Makhmudova U, Steinhagen-Thiessen E, Volpe M, Landmesser U. Advances in nucleic acid-targeted therapies for cardiovascular disease prevention. Cardiovasc Res 2024; 120:1107-1125. [PMID: 38970537 DOI: 10.1093/cvr/cvae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/14/2024] [Accepted: 05/03/2024] [Indexed: 07/08/2024] Open
Abstract
Nucleic acid-based therapies are being rapidly developed for prevention and management of cardiovascular diseases (CVD). Remarkable advancements have been achieved in the delivery, safety, and effectiveness of these therapeutics in the past decade. These therapies can also modulate therapeutic targets that cannot be sufficiently addressed using traditional drugs or antibodies. Among the nucleic acid-targeted therapeutics under development for CVD prevention are RNA-targeted approaches, including antisense oligonucleotides (ASO), small interfering RNAs (siRNA), and novel genome editing techniques. Genetic studies have identified potential therapeutic targets that are suggested to play a causative role in development and progression of CVD. RNA- and DNA-targeted therapeutics can be particularly well delivered to the liver, where atherogenic lipoproteins and angiotensinogen (AGT) are produced. Current targets in lipid metabolism include proprotein convertase subtilisin/kexin type 9 (PCSK9), apolipoprotein A (ApoA), apolipoprotein C3 (ApoC3), angiopoietin-like 3 (ANGPTL3). Several large-scale clinical development programs for nucleic acid-targeted therapies in cardiovascular prevention are under way, which may also be attractive from a therapy adherence point of view, given the long action of these therapeutics. In addition to genome editing, the concept of gene transfer is presently under assessment in preclinical and clinical investigations as a potential approach for addressing low-density lipoprotein receptor deficiency. Furthermore, ongoing research is exploring the use of RNA-targeted therapies to treat arterial hypertension by reducing hepatic angiotensinogen (AGT) production. This review summarizes the rapid translation of siRNA and ASO therapeutics as well as gene editing into clinical studies to treat dyslipidemia and arterial hypertension for CVD prevention. It also outlines potential innovative therapeutic options that are likely relevant to the future of cardiovascular medicine.
Collapse
Affiliation(s)
- Umidakhon Makhmudova
- Department of Cardiology, Angiology, and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
- Department of Endocrinology and Metabolic Diseases, Charite Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, Rome 00189, Italy
- Cardiology Department, IRCCS San Raffaele Roma, Via di Valcannuta 250, Rome 00166, Italy
| | - Ulf Landmesser
- Department of Cardiology, Angiology, and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Hindenburgdamm 30, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik/Centrum, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Centre for Cardiovascular Research, DZHK, Partner Site Berlin, Germany
| |
Collapse
|
39
|
Zhang K, Shi Y, Jin Z, He J. Advances in tumor vascular growth inhibition. Clin Transl Oncol 2024; 26:2084-2096. [PMID: 38504070 DOI: 10.1007/s12094-024-03432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
Tumor growth and metastasis require neovascularization, which is dependent on a complex array of factors, such as the production of various pro-angiogenic factors by tumor cells, intercellular signaling, and stromal remodeling. The hypoxic, acidic tumor microenvironment is not only conducive to tumor cell proliferation, but also disrupts the equilibrium of angiogenic factors, leading to vascular heterogeneity, which further promotes tumor development and metastasis. Anti-angiogenic strategies to inhibit tumor angiogenesis has, therefore, become an important focus for anti-tumor therapy. The traditional approach involves the use of anti-angiogenic drugs to inhibit tumor neovascularization by targeting upstream and downstream angiogenesis-related pathways or pro-angiogenic factors, thereby inhibiting tumor growth and metastasis. This review explores the mechanisms involved in tumor angiogenesis and summarizes currently used anti-angiogenic drugs, including monoclonal antibody, and small-molecule inhibitors, as well as the progress and challenges associated with their use in anti-tumor therapy. It also outlines the opportunities and challenges of treating tumors using more advanced anti-angiogenic strategies, such as immunotherapy and nanomaterials.
Collapse
Affiliation(s)
- Keyong Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuanyuan Shi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ze Jin
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jian He
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
40
|
Miao Y, Fu C, Yu Z, Yu L, Tang Y, Wei M. Current status and trends in small nucleic acid drug development: Leading the future. Acta Pharm Sin B 2024; 14:3802-3817. [PMID: 39309508 PMCID: PMC11413693 DOI: 10.1016/j.apsb.2024.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/15/2024] [Accepted: 04/12/2024] [Indexed: 09/25/2024] Open
Abstract
Small nucleic acid drugs, composed of nucleotides, represent a novel class of pharmaceuticals that differ significantly from conventional small molecule and antibody-based therapeutics. These agents function by selectively targeting specific genes or their corresponding messenger RNAs (mRNAs), further modulating gene expression and regulating translation-related processes. Prominent examples within this category include antisense oligonucleotides (ASO), small interfering RNAs (siRNAs), microRNAs (miRNAs), and aptamers. The emergence of small nucleic acid drugs as a focal point in contemporary biopharmaceutical research is attributed to their remarkable specificity, facile design, abbreviated development cycles, expansive target spectrum, and prolonged activity. Overcoming challenges such as poor stability, immunogenicity, and permeability issues have been addressed through the integration of chemical modifications and the development of drug delivery systems. This review provides an overview of the current status and prospective trends in small nucleic acid drug development. Commencing with a historical context, we introduce the primary classifications and mechanisms of small nucleic acid drugs. Subsequently, we delve into the advantages of the U.S. Food and Drug Administration (FDA) approved drugs and mainly discuss the challenges encountered during their development. Apart from researching chemical modification and delivery system that efficiently deliver and enrich small nucleic acid drugs to target tissues, promoting endosomal escape is a critical scientific question and important research direction in siRNA drug development. Future directions in this field will prioritize addressing these challenges to facilitate the clinical transformation of small nucleic acid drugs.
Collapse
Affiliation(s)
- Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
- Liaoning Medical Diagnosis and Treatment Center, Shenyang 110000, China
| | - Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yu Tang
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
- Liaoning Medical Diagnosis and Treatment Center, Shenyang 110000, China
| |
Collapse
|
41
|
Wang H, Fleishman JS, Cheng S, Wang W, Wu F, Wang Y, Wang Y. Epigenetic modification of ferroptosis by non-coding RNAs in cancer drug resistance. Mol Cancer 2024; 23:177. [PMID: 39192329 DOI: 10.1186/s12943-024-02088-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
The development of drug resistance remains a major challenge in cancer treatment. Ferroptosis, a unique type of regulated cell death, plays a pivotal role in inhibiting tumour growth, presenting new opportunities in treating chemotherapeutic resistance. Accumulating studies indicate that epigenetic modifications by non-coding RNAs (ncRNA) can determine cancer cell vulnerability to ferroptosis. In this review, we first summarize the role of chemotherapeutic resistance in cancer growth/development. Then, we summarize the core molecular mechanisms of ferroptosis, its upstream epigenetic regulation, and its downstream effects on chemotherapeutic resistance. Finally, we review recent advances in understanding how ncRNAs regulate ferroptosis and from such modulate chemotherapeutic resistance. This review aims to enhance general understanding of the ncRNA-mediated epigenetic regulatory mechanisms which modulate ferroptosis, highlighting the ncRNA-ferroptosis axis as a key druggable target in overcoming chemotherapeutic resistance.
Collapse
Affiliation(s)
- Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sihang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weixue Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Fan Wu
- Department of Hepatobiliary Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| | - Yu Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| |
Collapse
|
42
|
Çakan E, Lara OD, Szymanowska A, Bayraktar E, Chavez-Reyes A, Lopez-Berestein G, Amero P, Rodriguez-Aguayo C. Therapeutic Antisense Oligonucleotides in Oncology: From Bench to Bedside. Cancers (Basel) 2024; 16:2940. [PMID: 39272802 PMCID: PMC11394571 DOI: 10.3390/cancers16172940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/15/2024] Open
Abstract
Advancements in our comprehension of tumor biology and chemoresistance have spurred the development of treatments that precisely target specific molecules within the body. Despite the expanding landscape of therapeutic options, there persists a demand for innovative approaches to address unmet clinical needs. RNA therapeutics have emerged as a promising frontier in this realm, offering novel avenues for intervention such as RNA interference and the utilization of antisense oligonucleotides (ASOs). ASOs represent a versatile class of therapeutics capable of selectively targeting messenger RNAs (mRNAs) and silencing disease-associated proteins, thereby disrupting pathogenic processes at the molecular level. Recent advancements in chemical modification and carrier molecule design have significantly enhanced the stability, biodistribution, and intracellular uptake of ASOs, thereby bolstering their therapeutic potential. While ASO therapy holds promise across various disease domains, including oncology, coronary angioplasty, neurological disorders, viral, and parasitic diseases, our review manuscript focuses specifically on the application of ASOs in targeted cancer therapies. Through a comprehensive examination of the latest research findings and clinical developments, we delve into the intricacies of ASO-based approaches to cancer treatment, shedding light on their mechanisms of action, therapeutic efficacy, and prospects.
Collapse
Affiliation(s)
- Elif Çakan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Olivia D Lara
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Anna Szymanowska
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Emine Bayraktar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Medical Biology, Faculty of Medicine, University of Gaziantep, Gaziantep 27310, Turkey
| | | | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
43
|
Balcorta HV, Contreras Guerrero VG, Bisht D, Poon W. Nucleic Acid Delivery Nanotechnologies for In Vivo Cell Programming. ACS APPLIED BIO MATERIALS 2024; 7:5020-5036. [PMID: 38288693 DOI: 10.1021/acsabm.3c00886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
In medicine, it is desirable for clinicians to be able to restore function and imbue novel function into selected cells for therapy and disease prevention. Cells damaged by disease, injury, or aging could be programmed to restore normal or lost functions, such as retinal cells in inherited blindness and neuronal cells in Alzheimer's disease. Cells could also be genetically programmed with novel functions such as immune cells expressing synthetic chimeric antigen receptors for immunotherapy. Furthermore, knockdown or modification of risk factor proteins can mitigate disease development. Currently, nucleic acids are emerging as a versatile and potent therapeutic modality for achieving this cellular programming. In this review, we highlight the latest developments in nanobiomaterials-based nucleic acid therapeutics for cellular programming from a biomaterial design and delivery perspective and how to overcome barriers to their clinical translation to benefit patients.
Collapse
Affiliation(s)
- Hannia V Balcorta
- Department of Metallurgical, Materials, and Biomedical Engineering, College of Engineering, University of Texas at El Paso, 500 W. University Ave., El Paso, Texas 79968, United States
| | - Veronica G Contreras Guerrero
- Department of Metallurgical, Materials, and Biomedical Engineering, College of Engineering, University of Texas at El Paso, 500 W. University Ave., El Paso, Texas 79968, United States
| | - Deepali Bisht
- Department of Metallurgical, Materials, and Biomedical Engineering, College of Engineering, University of Texas at El Paso, 500 W. University Ave., El Paso, Texas 79968, United States
| | - Wilson Poon
- Department of Metallurgical, Materials, and Biomedical Engineering, College of Engineering, University of Texas at El Paso, 500 W. University Ave., El Paso, Texas 79968, United States
| |
Collapse
|
44
|
Subramanian G, Kalidasan K, Quah S, Han QCG, Chan J, Wacker MG, Sampath P. Breaking barriers: Innovative approaches for skin delivery of RNA therapeutics. Int J Pharm 2024; 661:124435. [PMID: 38986965 DOI: 10.1016/j.ijpharm.2024.124435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
RNA therapeutics represent a rapidly expanding platform with game-changing prospects in personalized medicine. The disruptive potential of this technology will overhaul the standard of care with reference to both primary and specialty care. To date, RNA therapeutics have mostly been delivered parenterally via injection, but topical administration followed by intradermal or transdermal delivery represents an attractive method that is convenient to patients and minimally invasive. The skin barrier, particularly the lipid-rich stratum corneum, presents a significant hurdle to the uptake of large, charged oligonucleotide drugs. Therapeutic oligonucleotides need to be engineered for stability and specificity and formulated with state-of-the-art delivery strategies for efficient uptake. This review will cover various passive and active strategies deployed to enhance permeation through the stratum corneum and achieve effective delivery of RNA therapeutics to treat both local skin disorders and systemic diseases. Some strategies to achieve selectivity between local and systemic administration will also be discussed.
Collapse
Affiliation(s)
- Gowtham Subramanian
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
| | - Kamaladasan Kalidasan
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
| | - Shan Quah
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
| | - Qi Chou Gavin Han
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore (NUS), 4 Science Drive 2, Singapore 117544, Singapore
| | - Justin Chan
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
| | - Matthias G Wacker
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore (NUS), 4 Science Drive 2, Singapore 117544, Singapore.
| | - Prabha Sampath
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore; Skin Research Institute of Singapore (SRIS), 11 Mandalay Road #17-01 Clinical Sciences Building, Singapore 308232, Singapore; Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Singapore; Program in Cancer & Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore.
| |
Collapse
|
45
|
Murphy AJ, Wilton SD, Aung-Htut MT, McIntosh CS. Down syndrome and DYRK1A overexpression: relationships and future therapeutic directions. Front Mol Neurosci 2024; 17:1391564. [PMID: 39114642 PMCID: PMC11303307 DOI: 10.3389/fnmol.2024.1391564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Down syndrome is a genetic-based disorder that results from the triplication of chromosome 21, leading to an overexpression of many triplicated genes, including the gene encoding Dual-Specificity Tyrosine Phosphorylation-Regulated Kinase 1A (DYRK1A). This protein has been observed to regulate numerous cellular processes, including cell proliferation, cell functioning, differentiation, and apoptosis. Consequently, an overexpression of DYRK1A has been reported to result in cognitive impairment, a key phenotype of individuals with Down syndrome. Therefore, downregulating DYRK1A has been explored as a potential therapeutic strategy for Down syndrome, with promising results observed from in vivo mouse models and human clinical trials that administered epigallocatechin gallate. Current DYRK1A inhibitors target the protein function directly, which tends to exhibit low specificity and selectivity, making them unfeasible for clinical or research purposes. On the other hand, antisense oligonucleotides (ASOs) offer a more selective therapeutic strategy to downregulate DYRK1A expression at the gene transcript level. Advances in ASO research have led to the discovery of numerous chemical modifications that increase ASO potency, specificity, and stability. Recently, several ASOs have been approved by the U.S. Food and Drug Administration to address neuromuscular and neurological conditions, laying the foundation for future ASO therapeutics. The limitations of ASOs, including their high production cost and difficulty delivering to target tissues can be overcome by further advances in ASO design. DYRK1A targeted ASOs could be a viable therapeutic approach to improve the quality of life for individuals with Down syndrome and their families.
Collapse
Affiliation(s)
- Aidan J. Murphy
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - May T. Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Craig S. McIntosh
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
46
|
Sekhar A, Kuttan A, Lange RA. Recent updates on therapeutic targeting of lipoprotein(a) with RNA interference. Curr Opin Cardiol 2024; 39:292-299. [PMID: 38547148 DOI: 10.1097/hco.0000000000001144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
PURPOSE OF REVIEW RNA interference (RNAi)-based therapies that target specific gene products have impacted clinical medicine with 16 FDA approved drugs. RNAi therapy focused on reducing plasma lipoprotein(a) [Lp(a)] levels are under evaluation. RECENT FINDINGS RNAi-based therapies have made significant progress over the past 2 decades and currently consist of antisense oligonucleotides (ASO) and small interfering RNA (siRNA). Chemical modification of the RNA backbone and conjugation of siRNA enables efficient gene silencing in hepatocytes allowing development of effective cholesterol lowering therapies. Multiple lines of evidence suggest a causative role for Lp(a) in atherosclerotic cardiovascular disease, and recent analyses indicate that Lp(a) is more atherogenic than low density lipoprotein- cholesterol (LDL-C). These findings have led to the 'Lp(a) hypothesis' that lowering Lp(a) may significantly improve cardiovascular outcomes. Four RNAi-based drugs have completed early phase clinical trials demonstrating >80% reduction in plasma Lp(a) levels. Phase 3 clinical trials examining clinical outcomes with these agents are currently underway. SUMMARY Currently, four RNAi-based drugs have been shown to be effective in significantly lowering plasma Lp(a) levels. Clinical outcome data from phase 3 trials will evaluate the Lp(a) hypothesis.
Collapse
Affiliation(s)
- Aravind Sekhar
- Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | | | - Richard A Lange
- Texas Tech University Health Sciences Center, El Paso, Texas, USA
| |
Collapse
|
47
|
Arjunan P, Kathirvelu D, Mahalingam G, Goel AK, Zacharaiah UG, Srivastava A, Marepally S. Lipid-nanoparticle-enabled nucleic acid therapeutics for liver disorders. Acta Pharm Sin B 2024; 14:2885-2900. [PMID: 39027251 PMCID: PMC11252464 DOI: 10.1016/j.apsb.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/08/2024] [Accepted: 03/19/2024] [Indexed: 07/20/2024] Open
Abstract
Inherited genetic disorders of the liver pose a significant public health burden. Liver transplantation is often limited by the availability of donor livers and the exorbitant costs of immunosuppressive therapy. To overcome these limitations, nucleic acid therapy provides a hopeful alternative that enables gene repair, gene supplementation, and gene silencing with suitable vectors. Though viral vectors are the most efficient and preferred for gene therapy, pre-existing immunity debilitating immune responses limit their use. As a potential alternative, lipid nanoparticle-mediated vectors are being explored to deliver multiple nucleic acid forms, including pDNA, mRNA, siRNA, and proteins. Herein, we discuss the broader applications of lipid nanoparticles, from protein replacement therapy to restoring the disease mechanism through nucleic acid delivery and gene editing, as well as multiple preclinical and clinical studies as a potential alternative to liver transplantation.
Collapse
Affiliation(s)
- Porkizhi Arjunan
- Center for Stem Cell Research (A Unit of inStem, Bengaluru), Christian Medical College Campus, Bagayam, Vellore 632002, Tamil Nadu, India
- Manipal academy for higher education, Mangalore 576104, Karnataka, India
| | - Durga Kathirvelu
- Center for Stem Cell Research (A Unit of inStem, Bengaluru), Christian Medical College Campus, Bagayam, Vellore 632002, Tamil Nadu, India
| | - Gokulnath Mahalingam
- Center for Stem Cell Research (A Unit of inStem, Bengaluru), Christian Medical College Campus, Bagayam, Vellore 632002, Tamil Nadu, India
| | - Ashish Kumar Goel
- Department of Hepatology, Christian Medical College & Hospital, Vellore 632004, Tamil Nadu, India
| | - Uday George Zacharaiah
- Department of Hepatology, Christian Medical College & Hospital, Vellore 632004, Tamil Nadu, India
| | - Alok Srivastava
- Center for Stem Cell Research (A Unit of inStem, Bengaluru), Christian Medical College Campus, Bagayam, Vellore 632002, Tamil Nadu, India
- Department of Hematology, Christian Medical College & Hospital, Vellore 632004, Tamil Nadu, India
| | - Srujan Marepally
- Center for Stem Cell Research (A Unit of inStem, Bengaluru), Christian Medical College Campus, Bagayam, Vellore 632002, Tamil Nadu, India
| |
Collapse
|
48
|
Tsamoulis D, Rallidis LS, Kosmas CE. Inclisiran: the preclinical discovery and development of a novel therapy for the treatment of atherosclerosis. Expert Opin Drug Discov 2024; 19:773-782. [PMID: 38804735 DOI: 10.1080/17460441.2024.2360415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of global morbidity and mortality. Lipid lowering therapy (LLT) constitutes the cornerstone of ASCVD prevention and treatment. However, several patients fail to achieve therapeutic goals due to low treatment adherence or limitations of standard-of-care (SoC) LLTs. Inclisiran represents a pivotal low-density lipoprotein cholesterol (LDL-C) lowering agent aiming to address current unmet needs in LLT. It is the first available small interfering RNA (siRNA) LLT, specifically targeting PCSK9 mRNA and leading to post-transcriptional gene silencing (PTGS) of the PCSK9 gene. AREAS COVERED Promising phase III trials revealed an ~ 50% reduction in LDL-C levels with subcutaneous inclisiran administration on days 1 and 90, followed by semiannual booster shots. Coupled with inclisiran's favorable safety profile, these findings led to its approval by both the EMA and FDA. Herein, the authors highlight the preclinical discovery and development of this agent and provide the reader with their expert perspectives. EXPERT OPINION The evolution of gene-silencing treatments offers new perspectives in therapeutics. Inclisiran appears to have the potential to revolutionize ASCVD prevention and treatment, benefiting millions of patients. Ensuring widespread availability of Inclisiran, as well as managing additional healthcare costs that may arise, should be of paramount importance.
Collapse
Affiliation(s)
- Donatos Tsamoulis
- Second Department of Cardiology, National & Kapodistrian University of Athens, Athens, Greece
| | | | - Constantine E Kosmas
- Second Department of Cardiology, National & Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
49
|
Yuan W, Shi X, Lee LTO. RNA therapeutics in targeting G protein-coupled receptors: Recent advances and challenges. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102195. [PMID: 38741614 PMCID: PMC11089380 DOI: 10.1016/j.omtn.2024.102195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
G protein-coupled receptors (GPCRs) are the major targets of existing drugs for a plethora of human diseases and dominate the pharmaceutical market. However, over 50% of the GPCRs remain undruggable. To pursue a breakthrough and overcome this situation, there is significant clinical research for developing RNA-based drugs specifically targeting GPCRs, but none has been approved so far. RNA therapeutics represent a unique and promising approach to selectively targeting previously undruggable targets, including undruggable GPCRs. However, the development of RNA therapeutics faces significant challenges in areas of RNA stability and efficient in vivo delivery. This review presents an overview of the advances in RNA therapeutics and the diverse types of nanoparticle RNA delivery systems. It also describes the potential applications of GPCR-targeted RNA drugs for various human diseases.
Collapse
Affiliation(s)
- Wanjun Yuan
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, People’s Republic of China
| | - Leo Tsz On Lee
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa 999078, Macau, China
| |
Collapse
|
50
|
Carlesso A, Hörberg J, Deganutti G, Reymer A, Matsson P. Structural dynamics of therapeutic nucleic acids with phosphorothioate backbone modifications. NAR Genom Bioinform 2024; 6:lqae058. [PMID: 38800826 PMCID: PMC11127634 DOI: 10.1093/nargab/lqae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 01/24/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Antisense oligonucleotides (ASOs) offer ground-breaking possibilities for selective pharmacological intervention for any gene product-related disease. Therapeutic ASOs contain extensive chemical modifications that improve stability to enzymatic cleavage and modulate binding affinity relative to natural RNA/DNA. Molecular dynamics (MD) simulation can provide valuable insights into how such modifications affect ASO conformational sampling and target binding. However, force field parameters for chemically modified nucleic acids (NAs) are still underdeveloped. To bridge this gap, we developed parameters to allow simulations of ASOs with the widely applied phosphorothioate (PS) backbone modification, and validated these in extensive all-atom MD simulations of relevant PS-modified NA systems representing B-DNA, RNA, and DNA/RNA hybrid duplex structures. Compared to the corresponding natural NAs, single PS substitutions had marginal effects on the ordered DNA/RNA duplex, whereas substantial effects of phosphorothioation were observed in single-stranded RNA and B-DNA, corroborated by the experimentally derived structure data. We find that PS-modified NAs shift between high and low twist states, which could affect target recognition and protein interactions for phosphorothioated oligonucleotides. Furthermore, conformational sampling was markedly altered in the PS-modified ssRNA system compared to that of the natural oligonucleotide, indicating sequence-dependent effects on conformational preference that may in turn influence duplex formation.
Collapse
Affiliation(s)
- Antonio Carlesso
- Department of Pharmacology, Sahlgrenska Academy, University of Gothenburg, Box 431, SE-405 30 Gothenburg, Sweden
| | - Johanna Hörberg
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 462, SE-405 30 Gothenburg, Sweden
| | | | - Anna Reymer
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 462, SE-405 30 Gothenburg, Sweden
| | - Pär Matsson
- Department of Pharmacology, Sahlgrenska Academy, University of Gothenburg, Box 431, SE-405 30 Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Sweden
| |
Collapse
|